Role of Platelets in Homing of Endothelial Progenitor Cells to the Site of Vascular Injury.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3681-3681
Author(s):  
Eli I. Lev ◽  
Zeev Estrov ◽  
Khatira Aboulfatova ◽  
David Harris ◽  
Juan F. Granada ◽  
...  

Abstract Endothelial Progenitor Cells (EPCs) are undifferentiated bone marrow cells that are rapidly mobilized upon vascular or tissue injury. The circulating EPCs home to and differentiate at the site of vessel injury to enhance repair of damaged endothelium. Much has been learnt of how EPCs adhere to and transmigrate on endothelial cells during angiogenesis, but little is know about how EPCs home to the site where endothelial cells are denuded and subendothelium is exposed following vascular injury. We hypothesized that platelets mediate EPC homing because they are the first cells that tether and adhere to subendothelium exposed by vascular injury. To test this hypothesis, we examined the platelet-EPC interaction under static and flow conditions. Human EPCs were purified from buffy coats by cell sorting using CD133 and VEGFR-2 as the cell markers. The EPCs were then incubated with washed platelets that were either at resting state or activated by 5 mg/ml of collagen. EPC-platelet interaction was measured in the form of heterotypic aggregation between two types of cells by flow cytometry (dual labeled with FITC-CD42a and PE-CD133). We found the EPCs attached to resting platelets, however, the percentage of platelets binding to EPCs significantly increased when platelets were activated by collagen (1.02% vs. 2.76%, p < 0.05). EPC-platelet aggregation was similarly detected in whole blood and platelet-rich plasma. When perfused over a monolayer of activated platelets under a shear stress of 2.5 dyn/cm2, the CD133 positive EPCs tethered to the platelet monolayer. The tethered EPCs either firmly adhered immediately or rolled a short distance before becoming adherent. This EPC-platelet interaction was calcium-dependent and inhibited by up to 60±11.5% by a polyclonal P-selectin antibody. In comparison, the monoclonal GP Iba antibody AK2, which blocks GP Ib-VWF interaction, had no effect. These results demonstrate for the first time a significant interaction between EPCs and activated platelets both at static and flow conditions. Our findings suggest that platelets may function as carrier cells that direct EPCs to the site of vascular injury. In addition to homing, platelets, which release several growth factors known to affect EPCs, may provide a supporting matrix on which adherent EPCs proliferate and differentiate.

2006 ◽  
Vol 96 (10) ◽  
pp. 498-504 ◽  
Author(s):  
Eli Lev ◽  
Zeev Estrov ◽  
Khatira Aboulfatova ◽  
David Harris ◽  
Juan Granada ◽  
...  

SummaryEndothelial progenitor cells (EPCs) mobilize from the bone marrow in response to tissue injury and participate in vascular repair. However, there is limited data about the homing mechanisms of EPCs to vascular injury sites. Recently animal experiments indicated that platelets playa role in recruitment of EPCs to injury sites. However, data on the possible interaction between platelets and EPCs within the human system are limited. We, therefore, examined in-vitro human platelet-EPC interaction under static and flow conditions. Human EPCs were isolated from donated buffy coats by magnetic microbeads and flow cytometry cell sorting using CD133 and VEGFR-2, respectively, as markers. Platelets were tested in the form of washed platelets, platelet rich plasma or whole blood. EPCs formed heterotypic aggregates with resting platelets under static conditions, an interaction that was greatly enhanced when platelets were activated by collagen, ADP or thrombin-activation peptide. The platelet-EPC interaction was inhibited by antibodies to P-selectin or P-selectin glycoprotein ligand-1 (PSGL-1), but not by antibodies to glycoproteins Ib-IX-V or IIb/IIIa. When perfused over activated platelets under shear stress of 2.5 dyn/cm2, EPCs tethered to platelayers and either adhered immediately or rolled a short distance before adhering. In addition, platelets promoted the colonization of adherent EPCs in culture conditions. Consistent with recent animal studies, these findings demonstrate that human EPCs interact in vitro with activated platelets under static and flow conditions, mediated through P-selectin–PSGL-1 interaction. This interaction may be a central mechanism for homing of EPCs to vascular injury sites.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3805-3805
Author(s):  
Charlotte Ann Bradbury ◽  
Tracey Buckley ◽  
Kate Fletcher ◽  
Peter Rose ◽  
David Fitzmaurice

Abstract Introduction: Endothelial progenitor cells (EPCs) are multipotent progenitor cells mainly residing in human bone marrow. In response to stimuli such as arterial ischaemia or venous thrombosis, EPCs are mobilised from the bone marrow, released into the circulation and home to sites of vascular injury where they promote endothelial regeneration, revascularization, vasoactive and angiogenic factor secretion, proteinase generation and vein wall remodelling. There is significant research interest into the potential therapeutic role of EPCs in vascular injury. However, to our knowledge there is no published data on the measurement of EPCs in a larger population of patients with comparison to thrombotic outcomes and other biomarkers known to be predictive of recurrence risk for example, D Dimer. Methods: The EXACT study is a randomised controlled trial of extended warfarin treatment versus observation only for the prevention of recurrent venous thromboembolism (VTE) and post thrombotic syndrome (PTS) in patients with an unprovoked VTE. Patients recruited to this study had blood samples taken 3 months post VTE event whilst still on warfarin prior to stopping or continuing anticoagulation according to their randomisation group. All patients were clinically followed up for 2 years following randomisation and recurrent thrombotic events (confirmed by imaging) were recorded. The blood samples were processed for Full Blood Count, INR, D Dimers and quantitation of Endothelial Cells by flow cytometry (circulating mature and progenitor). Endothelial cells were identified by low side scatter, weak CD45 (unlike haematopoetic cells) and expression of endothelial markers (CD146, VEGFR, VEGFR-2). Endothelial progenitor cells also demonstrated at least one marker of immaturity (CD34 and CD133). Apoptotic or dead cells were detected and excluded using 7-amino-actinomycin D staining. The number of EPC and circulating endothelial cell (CEC) events was divided by the number of CD45 positive events and multiplied by the total white count, which will give a value x103/microlitre. Results: At 3 months of treatment with warfarin, in 193 patients, samples of sufficient quality were processed by flow cytometry for EPC quantitation. In 182 of these patients, a sufficiently full citrated sample was also available for D Dimer quantitation. High D Dimer levels were associated with low EPC levels and visa versa (fig 1). Out of the 193 patients, during 2 year follow up, 19 patients (10%) went on to have a recurrent VTE. D Dimers at 3 months treatment were not significantly different in patients who had a recurrence versus those who didn't (Figure 2). There was also no significant difference in circulating mature endothelial cells (CEC) and total WCC (table 2). However, EPC levels were significantly lower in patients who went on to develop a thrombosis vs those who didn't (p<0.05) figure 3. Discussion: According to current guidelines, patients with unprovoked VTE are considered for long term anticoagulation following a discussion with the patient regarding VTE recurrence risk off treatment vs bleeding risk on treatment. D Dimer results can be helpful to refine the estimated recurrence risk (e.g. using the DASH2 score) but testing is only validated off anticoagulation (for at least 7 days) which can be cumbersome to organise. This study was unable to support the predictive value of D Dimers taken from patients on warfarin. If EPC quantitation from blood samples of patients is confirmed to be predictive of VTE recurrence this could represent a new informative biomarker to aid decision making without the need to interrupt anticoagulation therapy. It also has the potential to provide complimentary information. For example, in defining a truly "low risk" group in those with negative D Dimers after completion of treatment. Further larger studies are needed to confirm the predictive value of EPC quantitation for VTE recurrence in patient groups continuing or discontinuing anticoagulation (this study included both groups) and to confirm whether the same results apply to patients on DOACs. Disclosures No relevant conflicts of interest to declare.


2005 ◽  
Vol 94 (12) ◽  
pp. 1270-1279 ◽  
Author(s):  
Bruno Delorme ◽  
Agnès Basire ◽  
Carla Gentile ◽  
Florence Sabatier ◽  
Frédéric Monsonis ◽  
...  

SummaryCD146 is an adhesion molecule present on endothelial cells throughout the vascular tree. CD146 is also expressed by circulating endothelial cells (CECs) widely considered to be mature endothelial cells detached from injured vessels. The discovery of circulating endothelial progenitor cells (EPCs) originating from bone marrow prompted us to investigate whether CD146 circulating cells could also contains EPCs. We tested this hypothesis using an approach combining elimination of CECs by an adhesion step, followed by immunomagnetic sorting of remaining CD146+ cells from the non adherent fraction of cord blood mononuclear cells. When cultured under endothelial-promoting conditions, these cells differentiated as late outgrowth endothelial colonies: they grew as a cobblestone monolayer, were uniformly positive for endothelial markers and did not express leukocyte antigens. They highly proliferated and were expanded in long-term culture without alterations of their phenotypic and functional properties (DiI-ac-LDL uptake, wound repair, capillary-like network formation, and TNFα response). Moreover, these cells colonized a Matrigel plug in immunodeficient mice (NOD/SCID). Finally, using 4-color flow cytometry analysis of purified CD34+ cells, we clearly discriminated, CD146+ EPCs (CD146+ CD34+ CD45+ CD133+ or CD117+), and CD146+ CECs (CD146+ CD34+, CD45− CD133− or CD117−), both in cord and adult peripheral blood. The relative proportions of the two CD146+ subsets varied in patients with myocardial infarction as compared to healthy subjects. Our study establishes that, beside CECs, CD146+ circulating cells contain a subpopulation of EPCs with potential use in proangiogenic therapy. In addition, the dual measurement of CD146+ CECs and CD146+ EPCs offers a promising tool for monitoring vascular injury/regeneration processes in clinical situations.


2010 ◽  
Vol 88 (1) ◽  
pp. 121-129 ◽  
Author(s):  
Juliane Ladhoff ◽  
Bernhard Fleischer ◽  
Yoshiaki Hara ◽  
Hans-Dieter Volk ◽  
Martina Seifert

Author(s):  
T. Bulduk ◽  
A. U. Yalcin ◽  
O. M. Akay ◽  
S. G. Ozkurt ◽  
H. U. Teke ◽  
...  

Anemia is a common complication of chronic kidney disease (CKD). The most common cause of anemia in CKD is erythropoietin deficiency; and the most important cause of mortality in CKD patients is atherosclerotic vascular complications which are associated with endothelial damage. One of the methods evaluating vascular integrity is the cytometric measurement of circulating endothelial cells and endothelial progenitor cells in peripheral blood. The study aimed to investigate the effects of erythropoietin therapy on endothelial dysfunction by evaluating circulating endothelial cells and endothelial progenitor cells in peripheral blood using the technique of flow cytometry. Methods. A total of 55 hemodialysis patients were evaluated in three groups; those having erythropoietin therapy for at least last 3 months (n = 20) / not having erythropoietin for at least the last 3 months (n = 20) and the patients who started erythropoietin treatment during the study (n = 5). The control group consisted of 20 people. Blood values of the 3rd Group were investigated three times as baseline, 2nd week and 8th week CD34 +, CD105 + cells were evaluated as activated circulating endothelial cells; CD133 +, CD146 + cells were evaluated as activated endothelial progenitor cells. Results. There was no difference between the patients and healthy individuals in terms of circulating endothelial cells and endothelial progenitor cells. In the third group, no differences were observed in circulating endothelial cells / endothelial progenitor cell levels at baseline / 2nd and 8th weeks. There was no correlation between erythropoietin and circulating endothelial cells / endothelial progenitor cells. Conclusion. A correlation is not available between the therapeutic doses of erythropoietin used in hemodialysis patients and circulating endothelial cells / endothelial progenitor cell levels; supratherapeutic doses could change the results.


Sign in / Sign up

Export Citation Format

Share Document